1
|
Kessler JE, Park KN, Grizzle AJ, Hurwitz JT. Cost of illness of stage IV non-small cell lung cancer (NSCLC) positive for programmed cell death ligand 1 (PD-L1) in the US. Expert Rev Pharmacoecon Outcomes Res 2023; 23:55-61. [PMID: 36288082 DOI: 10.1080/14737167.2023.2140141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) positive for programmed cell death ligand 1 (PD-L1) may be eligible for targeted immunotherapies. Literature does not currently estimate direct costs associated with this population. We aimed to identify the total direct costs associated with PD-L1 positive stage IV NSCLC treated with immunotherapy. METHODS Using progression-free survival, overall survival, treatment-related serious adverse events leading to hospitalization, and end-of-life resource use, we estimated costs for one year of treatment in this incidence-based study. Data were obtained from online databases, guideline recommendations, clinical trials, and proprietary market share data. We summed the costs of PD-L1 immunohistochemistry (IHC) tests, drugs, hospitalizations, and deaths associated with treatment, estimating the overall cost-of-illness for stage IV NSCLC in the United States in 2021. RESULTS An estimated 22,711 patients in the US had stage IV NSCLC treated with PD-L1 immunotherapy in 2021. Total 2021 costs were estimated at $3.01 billion. Drugs (including immunotherapy, second-line chemotherapy, and other oncology drugs) accounted for nearly 97% ($2.91 billion) of the total. CONCLUSIONS PD-L1 positive stage IV NSCLC treatment is a costly condition with annual direct medical costs of $3.01 billion. The primary cost driver was immunotherapy, making up 74.6% of the total cost.
Collapse
Affiliation(s)
- Joshua E Kessler
- University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ, USA
| | - Kwon N Park
- University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ, USA
| | - Amy J Grizzle
- University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ, USA.,University of Arizona Center for Health Outcomes and PharmacoEconomic Research, Tucson, AZ, USA
| | - Jason T Hurwitz
- University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ, USA.,University of Arizona Center for Health Outcomes and PharmacoEconomic Research, Tucson, AZ, USA
| |
Collapse
|
2
|
Catalano M, Shabani S, Venturini J, Ottanelli C, Voltolini L, Roviello G. Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers (Basel) 2022; 14:6145. [PMID: 36551630 PMCID: PMC9777293 DOI: 10.3390/cancers14246145] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy is an ever-expanding field in lung cancer treatment research. Over the past two decades, there has been significant progress in identifying immunotherapy targets and creating specific therapeutic agents, leading to a major paradigm shift in lung cancer treatment. However, despite the great success achieved with programmed death protein 1/ligand 1 (PD-1/PD-L1) monoclonal antibodies and with anti-PD-1/PD-L1 plus anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4), only a minority of lung cancer patients respond to treatment, and of these many subsequently experience disease progression. In addition, immune-related adverse events sometimes can be life-threatening, especially when anti-CTLA-4 and anti-PD-1 are used in combination. All of this prompted researchers to identify novel immune checkpoints targets to overcome these limitations. Lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin (Ig) and Immunoreceptor Tyrosine-Based Inhibitory Motif (ITIM) domain (TIGIT), T cell immunoglobulin and mucin-domain containing-3 (TIM-3) are promising molecules now under investigation. This review aims to outline the current role of immunotherapy in lung cancer and to examine efficacy and future applications of the new immune regulating molecules.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Sonia Shabani
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Jacopo Venturini
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Carlotta Ottanelli
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Luca Voltolini
- Thoraco-Pulmonary Surgery Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Giandomenico Roviello
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
3
|
Barbar J, Armach M, Hodroj MH, Assi S, El Nakib C, Chamseddine N, Assi HI. Emerging genetic biomarkers in lung adenocarcinoma. SAGE Open Med 2022; 10:20503121221132352. [PMID: 36277445 PMCID: PMC9583216 DOI: 10.1177/20503121221132352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Comprehensive genomic profiling is a next-generation sequencing approach used to
detect several known and emerging genomic alterations. Many genomic variants
detected by comprehensive genomic profiling have become recognized as
significant cancer biomarkers, leading to the development of major clinical
trials. Lung adenocarcinoma has become one of the most targeted cancers for
genomic profiling with a series of actionable mutations such as EGFR, KRAS,
HER2, BRAF, FGFR, MET, ALK, and many others. The importance of these mutations
lies in establishing targeted therapies that significantly change the outcome in
lung adenocarcinoma besides the prognostic value of some mutations. This review
sheds light on the development of the comprehensive genomic profiling field,
mainly lung adenocarcinoma, and discusses the role of a group of mutations in
this disease.
Collapse
Affiliation(s)
- Jawad Barbar
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Maria Armach
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammad Hassan Hodroj
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Sahar Assi
- Department of Internal Medicine,
American University of Beirut Medical Center, Beirut, Lebanon
| | - Clara El Nakib
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Nathalie Chamseddine
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Hazem I Assi
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon,Hazem I Assi, Department of Internal
Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute,
American University of Beirut Medical Center, P.O. Box: 11-0236, Riad El Solh,
Beirut 1107 2020, Lebanon.
| |
Collapse
|
4
|
Lê H, Seitlinger J, Lindner V, Olland A, Falcoz PE, Benkirane-Jessel N, Quéméneur E. Patient-Derived Lung Tumoroids—An Emerging Technology in Drug Development and Precision Medicine. Biomedicines 2022; 10:biomedicines10071677. [PMID: 35884982 PMCID: PMC9312903 DOI: 10.3390/biomedicines10071677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/16/2022] Open
Abstract
Synthetic 3D multicellular systems derived from patient tumors, or tumoroids, have been developed to complete the cancer research arsenal and overcome the limits of current preclinical models. They aim to represent the molecular and structural heterogeneity of the tumor micro-environment, and its complex network of interactions, with greater accuracy. They are more predictive of clinical outcomes, of adverse events, and of resistance mechanisms. Thus, they increase the success rate of drug development, and help clinicians in their decision-making process. Lung cancer remains amongst the deadliest of diseases, and still requires intensive research. In this review, we analyze the merits and drawbacks of the current preclinical models used in lung cancer research, and the position of tumoroids. The introduction of immune cells and healthy regulatory cells in autologous tumoroid models has enabled their application to most recent therapeutic concepts. The possibility of deriving tumoroids from primary tumors within reasonable time has opened a direct approach to patient-specific features, supporting their future role in precision medicine.
Collapse
Affiliation(s)
- Hélène Lê
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Transgène SA, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
| | - Joseph Seitlinger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Véronique Lindner
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Pierre-Emmanuel Falcoz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (H.L.); (J.S.); (V.L.); (A.O.); (P.-E.F.); (N.B.-J.)
- Faculty of Medicine and Pharmacy, University Hospital Strasbourg, 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Eric Quéméneur
- Transgène SA, 400 Boulevard Gonthier d’Andernach, 67400 Illkirch-Graffenstaden, France
- Correspondence:
| |
Collapse
|